Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Pharmacol. Mar 9, 2015; 4(1): 58-74
Published online Mar 9, 2015. doi: 10.5497/wjp.v4.i1.58
Improving cancer therapy by targeting cancer stem cells: Directions, challenges, and clinical results
Pier Adelchi Ruffini, Valentina Vaja, Marcello Allegretti, Research and Development Department, Dompé Farmaceutici S.p.A., 20122 Milano, Italy
Author contributions: Ruffini PA, Vaja V and Allegretti M contributed to this paper.
Conflict-of-interest: Pier Adelchi Ruffini, Valentina Vaja and Marcello Allegretti are employees of Dompé Farmaceutici S.p.A.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Pier Adelchi Ruffini, MD, PhD, Research and Development Department, Dompé Farmaceutici S.p.A., via S. Lucia 6, 20122 Milano, Italy. pieradelchi.ruffini@dompe.com
Telephone: +39-02-58383500 Fax: +39-02-58383324
Received: September 30, 2014
Peer-review started: October 1, 2014
First decision: October 28, 2014
Revised: November 26, 2014
Accepted: February 9, 2015
Article in press: February 11, 2015
Published online: March 9, 2015

Abstract

Cancer stem cells (CSC) are a rare cell population within a tumor characterized by the ability to form tumors following injection into an immunocompromised host. While the role of CSC has been clearly established in animal models, evidence of their clinical relevance has been harder to demonstrate. A number of markers, or combination thereof, have been used to detect and measure, although non-specifically, CSC in almost all human tumors. Several pathways have been identified as crucial for, but not necessarily unique to, CSC survival and proliferation, and novel agents have been designed to target such pathways. A number of such agents have entered early phase development. Further, drugs that have long been marketed for non-oncological indications have been redirected to oncology as they appear to affect one or more of such pathways. This article aims to review the available evidence on the clinical relevance of CSC from a drug development standpoint and the results of early phase clinical trials of agents interfering with the above pathways. It also discusses limitations of current clinical trial design and endpoints to demonstrate anti-CSC activity as well as possible strategies to overcome these limitations.

Key Words: Cancer stem cells, Cancer, Time to new metastasis, Tumor heterogeneity, Drug development

Core tip: Cancer stem cells are a rare cell population with two embedded fundamental properties: self-renewal and differentiation into the heterogeneous lineages of cancer cells that comprise the tumor. While the identification and role of such cells has been clearly established in vitro and in animal models, uncertainty remains as to the role they play in human tumors and their relevance as therapeutic targets. The article summarizes and discusses the latest findings and challenges on cancer stem cell research, the significance of these cells beyond experimental models and suggests avenues for development of cancer stem cell targeting agents.



INTRODUCTION

In order to explain tumor heterogeneity, the clonal theory of cancer initiation and progression was proposed[1], stating that each cell within a tumor has equal potential in acquiring genetic and/or epigenetic changes, conferring growth advantages and generating new tumors. This model presumes that all cells within a given tumor have the same tumorigenic potential, and it is called classical or stochastic.

In the cancer stem cell paradigm of tumors, heterogeneous neoplastic cells are organized in a hierarchy where only a small subset of cancer cells, the cancer stem cells (CSC), is responsible for tumor initiation and growth maintenance. According to a consensus definition[2], a CSC is a cell within a tumor that possesses the capacity to self-renew and to generate the heterogeneous lineages of cancer cells that comprise the tumor. CSC could be tissue stem cells or a more differentiated progeny, which acquired self-renewal capacity[3-5]. There are 2 mechanisms that could mediate the transformation of normal stem cells to CSC[6,7]: (1) Early progenitor cells can gain mutations, which gives them self-renewal capacity; and (2) Fully differentiated cells or cells in the late progenitor stage can become de-differentiated to acquire the properties of stem cells.

Activation of oncogenes or inactivation of tumor suppressor genes are the major types of mutations involved in these processes. CSC may indeed arise from normal stem cells by mutation of genes that make the stem cells cancerous, but this may not be the case in all tumors. For example, in blast crisis chronic myelogenous leukemia (CML), a committed granulocyte-macrophage progenitor may acquire self-renewal capacity and thus “reacquire” stem-like properties due to the effects of later mutations[2]. It is conceivable that more differentiated cells can, through multiple mutagenic events, acquire the self-renewal capacity and immortality that typify CSC. In these examples, a differentiated cell, not the tissue stem cell, eventually evolves to become a full-blown CSC.

The first compelling evidence on the existence of CSC was presented by Bonnet et al[8] through their seminal work on leukemic stem cells, where they showed that these cells possess characteristic functional properties of stem cells. They demonstrated that the cell capable of initiating human acute myeloid leukemia in non-obese diabetic mice with severe combined immunodeficiency (SCID)-termed the SCID leukemia-initiating cell-possesses the differentiative and proliferative capacities and the potential for self-renewal expected of a leukemic stem cell. The SCID leukemia-initiating cells were able to differentiate in vivo into leukemic blasts, indicating that the leukemic clone is organized as a hierarchy.

CSC have been identified also in solid tumors, including breast, lung, colon, prostate and pancreatic cancers, all supporting the model that cancer derives from a subpopulation of CSC capable of self-renewal to initiate and sustain tumor growth[9,10].

Functional assays were performed in all of these studies, in which cell suspensions of cancer cells were inoculated into immunodeficient mice. This assay is currently considered the gold standard for studying the degree of stemness in a subpopulation of human cancer cells that drives tumorigenicity. In fact, CSC are not necessarily derived from stem cells although they share functional similarities to normal stem cells.

Therefore, the definition of CSC remains mainly operational, i.e., the ability to form tumors in an animal model, which is, although imperfect[11], regarded as the best functional assay to meet the two critical criteria of the consensus CSC definition[2]. The implementation of this approach explains the use of alternative terms in the literature, such as “tumor-initiating cell” and “tumorigenic cell” to describe putative CSC.

However, the results of this assay have been questioned, as it may measure the ability of human tumor cells to grow in immunodeficient mice and not necessarily the actual frequency of CSC in the tumor. It has been demonstrated that only rare human cancer cells (0.1%-0.0001%) form tumors when transplanted into immunodeficient mice. However, modifications to xenotransplantation assays (i.e., the degree of immunodeficiency of mice) can dramatically increase the detectable frequency of tumorigenic cells, demonstrating that they are common in some human cancers[12]. Furthermore, introducing human tumor cells in Matrigel containing a cocktail of growth factors to a foreign species such as a mouse may compromise the growth of human cancer cells[13], indicating the importance of the tumor microenvironment in tumorigenesis. Last, the nature of the proteolytic enzymes used, the duration of incubation, and the temperature at which the tumor cells are dissociated into single cells in order to perform xenotransplantation might further result in the under-estimation of the percentage of CSC, even with the same type of tumor. It follows that if the cancer initiating cells are not rare, the hierarchical model of CSC may be questioned[12]. It should be stressed that the aforementioned findings do not disprove the CSC paradigm. The CSC paradigm is directly related to tumor heterogeneity and tumor’s hierarchical organization, and not to the absolute number of these cells[14]. Thus, evidence of CSC in many human tumors has been described using the xenotransplantation model by highlighting that a fraction of tumor cells, and not all cancer cells, are tumorigenic and may thus represent a distinct therapeutic target[15].

From a therapeutic perspective, it is important to acknowledge that other targets exist beyond the CSC themselves to affect this cell population. In fact, the tumor microenvironment, the CSC niche, and cytokine loops play essential roles in the maintenance of CSC and in tumor growth and development[16,17]. For example, Feng et al[18] demonstrated that tumor microenvironment stromal cells create a growth advantage for CSC in a model of multiple myeloma. In that study, the proliferative capacity of multiple myeloma stem cells was stronger when grown in the presence of stromal cells from patients with myeloma than it was in the presence of stromal cells from normal control bone marrow.

If CSC are relatively refractory to therapies (e.g., chemotherapy, radiotherapy) that have been developed to eradicate the rapidly dividing cells that constitute the majority of the non-stem cell component of tumors, then such therapies are unlikely to be curative and relapses would be expected. If correct, the cancer stem cell hypothesis would require adjustments to the way tumors are diagnosed and treated. The objective would have to be not only to eliminate the bulk of rapidly dividing but terminally differentiated components of the tumor responsible for tumor-associated symptoms, but also the minority stem cell population that fuels tumor growth and is responsible for disease relapse and metastases.

CANCER STEM CELL SURFACE MARKERS

Besides discussion about the pitfalls of the functional assay used to identify CSC, it is intuitive that such an assay is unfit for clinical development of any novel drug as it would require ready availability of immunodeficient mice to be injected with single cell suspensions of patient-derived cancer cells before and after treatment, if one is to demonstrate presence of CSC and their reduction following treatment.

Therefore, more practical assays have been developed to be implemented in clinical research. An in vitro tumorsphere assay was used to evidence the self-renewal properties of CSC in human brain tumors[19,20]. This assay is now widely used in clinical research inasmuch as it requires live cells, and this requirement may not be always satisfied in large, multicenter clinical trials. The easiest way to identify and measure CSC within tumors or in the bloodstream would be to use CSC-specific or - associated cell surface marker proteins. By staining cells with antibodies against these markers, populations of interest could be easily identified and measured by either flow cytometry (which requires live cells) or immunohistochemistry (which does not require live cells).

Several markers are commonly used to isolate CSC in normal and tumor tissue, but it is not clear if they are universal markers for characterizing CSC from all tumor types. Furthermore, expression of markers may not be restricted to the CSC population and may be present in normal stem cells, early progenitor cells and normal tissues.

Although many markers have been proposed to define CSC populations, here we focus on the ones most widely used in the literature (Table 1).

Table 1 Commonly used Cancer Stem Cells Markers.
MarkerAssociation with chemoresistanceReported malignancies
CD34Hematological malignancies (in combination with CD38)[8,36]
CD38Hematological malignancies (in combination with CD34)[8,36]
CD24Yes (in combination with CD44)[63]Breast, prostate, pancreas (in combination with CD44)[15,23]
CD44Yes (in combination with CD24)[63]Breast, pancreas, stomach, prostate, head and neck, ovary, colon (in combination with CD24)[15,23]
CD133Yes[24,25]Brain, pancreas, lung, liver, prostate, stomach, colon, head and neck[15,23]
ALDHYes[60,61]Breast, lung, head and neck, colon, liver, ovary[15,26]

The pentaspan transmembrane glycoprotein CD133, also known as prominin-1, was discovered as the target of the AC133 monoclonal antibody, specific for the CD34+ population of hematopoietic stem cells. Subsequently, it was shown to be expressed by a number of normal stem and progenitor cells including those of the epithelium, where it is expressed on the apical surface[21]. The biological role of CD133 has yet to be clarified.

Best known for being expressed on the tumor-initiating population of brain neoplasms[22] the CD133+ phenotype has recently been used to define the CSC populations in lung, pancreatic, liver, prostate, gastric, colorectal, as well as head and neck cancers, and CD133+ cells clearly generated tumors in immunocompromised mice more efficiently than their CD133-counterpart[23]. Also, CD133+ CSC display resistance to traditional chemotherapies[24,25].

A valid stem cell marker among several malignant and non-malignant tissues is aldehyde dehydrogenase (ALDH). ALDH is an intracellular enzyme that oxidizes aldehydes. It holds the attractive distinction of being not only a potential marker of “stemness”, but it potentially plays a role in the biology of tumor initiating cells as well[26].

Traditional methods such as immunohistochemistry and blotting are used to detect levels of ALDH in tissues and cells, but the method of choice to measure ALDH activity is the flow cytometry-based Aldefluor assay[27]. In the past few years, Aldefluor has been used to characterize CSC in breast, lung, head and neck, colon, and liver tumors and cell lines. Although CSC are enriched in ALDH+ populations in several tissues, it is important to acknowledge possible limitations of this marker, especially when used as a single marker. Enzymatic activity measured by Aldefluor is much more transient than the expression of traditional surface markers and may be altered by treatment of tumors or cells[23]. ALDH activity has been used as a marker for CSC in malignancies characterized by the presence and/or absence of cell-surface markers like the CD44+/CD24-/low and CD133+ populations such as breast cancer and ovarian cancer, respectively[9,28] and early reports suggest that the use of ALDH may “refine” the CSC population because the CSC populations defined above and the ALDH+ populations rarely overlap completely[23].

CD44 is a transmembrane glycoprotein which mediates cell adhesion and migration by binding extracellular matrix components such as hyaluronic acid and osteopontin, or by activating receptor tyrosine kinases related to tumor progression, invasion and metastasis. CD44 is reported as a CSC marker across several tissues, including breast, pancreas, gastric, prostate, head and neck, ovarian, and colon[15]. In both preclinical models and clinical research, however, CD44 is most commonly used in combination with other markers, e.g., CD24.

CD24 is a glycosylphosphatidylinositol-linked cell surface protein expressed in various solid tumors[29]. Expression of CD24 in adult non-malignant tissue is limited to B cells, granulocytes, and epithelial cells[30,31]. The normal function of CD24 outside of B cells is poorly understood. The presence or absence of CD24 on the cell surface has been used as a marker for putative CSC. Breast and prostate CSC (CD44+/CD24-) were found to have increased adhesion, invasion and migration characteristics when compared with CD24-expressing cells[32], and prostate cells that were able to migrate through Matrigel had suppressed CD24 expression along with decreased levels of E-cadherin[33].

The presence or absence of CD24 as a CSC marker seems to be tissue specific, i.e., breast (CD44+/CD24-/low)[9], prostate (CD44+/CD24-)[34] and pancreatic (CD44+/CD24+/ESA+)[35] cancers.

All the above markers have been used alone or in combination to define CSC populations in solid tumors. The most commonly used markers to define CSC in hematological malignancies are CD34 and CD38, which are used in combination[8,36]. Also for these markers, expression is not restricted to CSC, as they are expressed also by normal hematopoietic and non-hematopoietic cells as well as hematopoietic stem and progenitor cells (CD34).

CLINICAL EVIDENCE FOR A ROLE OF CSC IN CANCER INITIATION, RELAPSE AND METASTASES

Following the demonstration that only CSC identified in patients’ tumor cells by the previously described markers can form tumors following injection into immunodeficient mice, a number of studies has been conducted to measure CSC in human tumors and to explore their association with prognosis. These studies are important as they would contribute to validate CSC as a therapeutic target and to confirm the correlation between the presence of such cells in tumor samples with unfavorable outcome, independent of any other prognostic factor. Methodologically, most of these studies are retrospective, which in itself carries limitations due to change over time in diagnostic and therapeutic procedures. Breast cancer is by far the most thoroughly investigated solid tumor. Two meta-analyses have been published. The first reported on 9 studies including a total of 2121 patients and demonstrated that the presence of ALDH+ CSC and CD44+/CD24-/low CSC was significantly associated with poor overall survival[37]. All of the studies included in the meta-analysis used either ALDH or CD44/CD24 as CSC marker. More recently, a second meta-analysis on 3274 patients using ALDH as the sole CSC marker (sharing 7 out of 16 studies and more than 2000 patients with the first meta-analysis) confirmed the association between the presence of ALDH+ CSC and reduced patient survival irrespective of the different cutoff values used in the different studies to define ALDH positivity[38]. However, breast cancer can be divided into several subtypes, either into histopathological categories based upon expression or lack of hormone receptors and HER2 amplification, or into molecularly identifiable categories using gene expression signatures[39]. The presence and clinical significance of CSC have been investigated in most subtypes[40,41]. In this respect, it is interesting to note that CSC have been shown to be represented at an increased percentage in some breast cancer subtypes. For example, Triple Negative Breast Cancer (TNBC) has been described as the most CSC enriched breast cancer subtype[42-44] Further molecular dissection of TNBC identified mesenchymal stem cell-Like[45] and claudin-low[46] as the most CSC enriched subgroups. It has been shown in vitro that breast cancer cell lines of the luminal subtype do not always contain a sizeable ALDH+ cell population, whereas 16/16 basal-like cell lines displayed a CSC population identified by the same marker[47]. Indeed, with a cutoff for positivity set at ≥ 5% in most studies, it would appear that a minority of tumors display a sizeable CSC population identified by a single marker or combination of markers (i.e., CD44+/CD24-, ALDH+)[37,38]. It remains to be demonstrated whether this is due to suboptimal markers to identify CSC and/or the limited overlap between CSC populations identified by either marker alone[41,48,49].

In other solid tumors similar results have been reported, although the number of patients studied is in general far lower. In ovarian cancer, the presence of ALDH+/CD133+ cells in debulked primary tumor specimens correlated with reduced disease-free and overall survival in 56 ovarian cancer patients[28]. These findings were confirmed by a meta-analysis of 1258 patients from 7 studies, in which ALDH was used as a single CSC marker[50]. In prostate cancer, detection of ALDH+ CSC correlated with shorter median survival in hormone naïve, but not in castration resistant, tumors. The two groups consisted of 100 and 107 patients, respectively[51]. In gastric cancer, presence of ALDH+ CSC was found to be an independent prognostic factor for both overall survival and recurrence-free survival[52] in a population of 216 patients. In pancreatic cancer, expression of the CSC markers CD133, CD44 and CD24 was found to correlate with poor prognosis in a sample of 65 patients[53]. In hematological malignancies, it has been found that CD34+/ALDH+ CSC in the bone marrow of 68 patients with acute myeloid leukemia at diagnosis correlated with reduced survival probability[54]. In conclusion, retrospective studies have found a negative prognostic role of pretreatment CSC, in keeping with their alleged resistance to chemo- and radiotherapy.

EVIDENCE OF CSC RESISTANCE TO CHEMOTHERAPY

Breast cancer is the solid tumor in which this issue has been explored most thoroughly. Breast cancer CSC have been shown to display resistance to commonly used chemotherapeutic agents (e.g., paclitaxel) in vitro[55] and in mice[56,57]. In patients, neoadjuvant (i.e., administered before surgery) chemotherapy represents the ideal setting for assessing the efficacy of chemotherapy against CSC, as the primary tumor is readily available to serial biopsies before, during and at the completion of treatment. In the majority of clinical trials of neoadjuvant chemotherapy, the proportion of CSC identified by marker expression and/or mammosphere forming efficiency was found to be increased following treatment[58-60] despite reduction of tumor size, highlighting that chemosensitivity of the bulk tumor cells is not shared by CSC.

In these studies, 56% or fewer patients had ALDH+ CSC in their tumors at diagnosis, using a cutoff of ≥ 5% to ≥ 20% (reviewed in[61]). In keeping with preclinical findings[41,62], it was reported that the ALDH+ phenotype[60,61], or the CD44+/CD24- phenotype[63], is associated with chemoresistance However, the studies by Lee et al[59] and Gong et al[62] came to the opposite conclusion, i.e., ALDH+ CSC correlated with increased pathologic or clinical responses, respectively. The reasons for this discrepancy could be different chemotherapy regimens administered to patients, different patient populations, or different antibody and cutoff values used to evaluate ALDH positivity in breast cancer samples.

The study by Alamgeer et al[61] is the only prospective evaluation of ALDH+ cells in serial biopsies of breast cancer. Biopsies were obtained before neoadjuvant chemotherapy, following 4 cycles of either one of two chemotherapy regimens, and at completion of treatment in 119 patients. Dynamic changes of ALDH were recorded, with patients switching from ALDH- to ALDH+ and vice versa in 27% and 19% of cases, respectively, at the biopsy after 4 cycles of chemotherapy. There are several possible explanations for these findings. First, there may be interconversion between the stem cell like and non-stem cell like phenotype, as previously described in preclinical models[64]. However, conversion from ALDH+ to ALDH- with chemotherapy alone (i.e., without a CSC targeting agent) is not in keeping with the alleged chemoresistance of CSC. Second, ALDH+ cells have been described to be localized at the center of the tumor mass, whereas CD44+/CD24- cells are localized at the edges[49]. Thus, serial biopsy of a tumor mass may lead to increase or decrease of either cell population depending on the area sampled. Third, for biopsies negative at baseline, which convert to positive after 4 wk of treatment, there might have been ALDH positivity below the 5% cutoff value of an otherwise positive baseline sample. However, the most intriguing finding of the study is that the disease-free survival of those patients who did not achieve pathologic complete response (pCR) but displayed elimination of ALDH+ cells in the primary tumor, is similar to that of patients experiencing pCR[61].

Another prospective study, the AVASTEM trial, was designed to evaluate whether the addition of bevacizumab to conventional chemotherapy alters the proportion of CSC in breast cancer patients receiving neoadjuvant treatment. It is an open label, randomized phase II trial in which tumor biopsies are required at baseline and after the 4 initial cycles of treatment to evaluate variations in the ALDH+ CSC population[65]. Preclinical work demonstrated that anti-angiogenic agents (e.g., bevacizumab) may increase breast cancer CSC via the generation of tumor hypoxia[66]. Indeed, CSC have been described to reside near hypoxic regions in solid tumors[67-70]. Results of AVASTEM will confirm or disprove these preclinical findings.

MOLECULAR SIGNALING PATHWAYS AS THERAPEUTIC TARGETS IN CSC

Signaling pathways are essential for normal stem cells with respect to self-renewal, proliferation and differentiation. Accumulating evidence suggests that maintenance of the CSC population in different human tumors employs the same signaling pathways as in normal stem cells, although an alteration of these pathways during the development of cancer has occurred, and this event has led to dysregulation of stem cell self-renewal and contributes to tumor proliferation[71,72]. Therefore, targeting these aberrant signaling pathways that are important for the formation of CSC seems to offer a new strategy for cancer therapy.

Most of these pathways have been described in dedicated reviews[15]. In this section, we will present and discuss clinical data concerning anti-tumor activity and side effects related to targeting of each pathway.

The hedgehog signaling pathway

The hedgehog (Hh) pathway plays a crucial role in development and patterning during mammalian embryogenesis, and it is essential for the maintenance of stem cells[73]. The Hh pathway is activated when Hh protein binds to and inhibits the transmembrane protein called Patched, leading through activation of the Smoothened function to the regulation of target genes involved in many cellular functions including metastasis, proliferation, survival, and pathway auto-regulation[74,75]. Deregulation of the Hh pathway is associated with numerous human malignancies and is a critical factor affecting the outcome of treating the disease[76]. Recently, it has been suggested that the Hh pathway is essential for the maintenance of CSC in various human cancers, including colorectal, pancreatic and gastric cancer[77,78], and it is also responsible for treatment resistance of cancer cells[79]. Thus, inhibitors that block any step of the Hh signaling pathway may result in reduction of CSC and overcoming treatment resistance.

One Hh inhibitor, vismodegib, is approved for treatment of metastatic or locally advanced, unresectable basal cell carcinoma. Vismodegib 150 mg/d administered orally produced a sizeable and lasting response rate, including complete responses, in patients with basal cell carcinoma, suggesting activity also on bulk tumor cells[80,81]. In a randomized trial of vismodegib vs placebo in patients with basal cell nevus syndrome, an autosomal dominant disorder causing a markedly increased incidence of basal cell carcinoma, medulloblastoma, and rhabdomyosarcoma, new basal cell carcinoma formation rate was decreased by vismodegib from 29/year to 2/year[82]. This tumor prevention capability would suggest suppression of CSC activity, however, no formal investigation of anti-CSC activity has been reported. Vismodegib administration may result in adverse reactions, so that in the latter trial, 54% of patients had to discontinue treatment due to adverse reactions[82], with 25% of patients experiencing serious adverse events in the phase II trial regarding basal cell carcinoma[81]. Ongoing and completed clinical trials using Smoothened antagonists were recently reviewed by Amakye et al[83] (Table 2).

Table 2 Pathways and drug candidates targeting cancer stem cells.
PathwayTargetsCompoundsClinical development
HedgehogSmoothened1Vismodegib[80-82]Medulloblastoma
Pancreatic cancer
Hematological malignancies
Notchγ-secretaseMK-0752[94]Breast cancer
DLL4Demcizumab[88-90]Pancreatic cancer
NSCLC
Ovarian cancer
MEDI0639Solid tumors
Wntβ-cateninVantictumab[101]Breast cancer
Pancreatic cancer
NSCLC
OMP-54F28[102]Ovarian cancer
Pancreatic cancer
Hepatic cancer
CXCR1/2-CXCL8CXCR1Reparixin[113,114]Breast cancer
SCH563705
CXCR1/2
IL-6/JAK/STAT3IL-62Tocilizumab
JAK1, JAK23Tofacitinib
4ChloroquineBreast cancer[57]
Notch signaling pathway

The Notch signaling pathway plays several crucial roles in the communication between cells[84,85]. The Notch pathway is activated through ligand-receptor interactions of four receptors (Notch-1-Notch-4) and five Notch ligands (Delta-like1, 3, 4 and Jagged1, 2)[85], resulting in the expression of multiple target genes. The Notch pathway is involved in stem cell proliferation, differentiation, and apoptosis, but its role in tumorigenesis can be either oncogenic or oncosuppressive[85]. In fact, Notch functions as an oncogenic protein in most human cancers including cervical, lung, colon, head and neck, prostate and pancreatic cancer, while it may act as a tumor suppressor in hepatocellular carcinoma, skin and small cell lung cancer[86,87].

The delta-like ligand 4 (DLL4) contributes to CSC self-renewal and vascular development and can bind to any of the 4 receptors. Two anti-DLL4 monoclonal antibodies are in development (Table 2). Demcizumab (OMP-21M18, Oncomed) is a humanized IgG2 antibody specific for DLL4 with as dual mechanism of action: targeting of cancer stem cells and inhibition of angiogenesis. It has been evaluated as a single agent in patients with advanced malignancies[88] and in two phase Ib dose escalation clinical trials for frontline treatment of metastatic pancreatic adenocarcinoma[89] and non-squamous non-small cell lung cancer[90] in combination with gemcitabine and carboplatin + pemetrexed, respectively. In pancreatic adenocarcinoma, median Progression-Free Survival observed did not differ significantly from what would be expected from single agent treatment with gemcitabine in the same patient population[89]. In order to comply with the new standard of care in this disease[91], patients are now being treated with demcizumab in combination with gemcitabine + nab-paclitaxel[89]. In non-squamous non-small cell lung cancer, median Progression-Free Survival was around 6 mo[90]. In both studies, the objective response rate was significant (25% and 46%, respectively), but the dual mechanism of action makes it difficult to disentangle effects on CSC from anti-angiogenic activity. The most common adverse reactions included nausea, fatigue and hypertension, the latter of which was managed with anti-hypertensives[88-90]. Also, late cardiopulmonary toxicity was recorded in a few patients treated for over 5 mo, leading to a shorter course of treatment and monitoring for early signs of toxicity by serial measurements of B-type natriuretic peptide levels[89,90].

MEDI0639 (medimmune) is an IgG1 lambda (IgG1λ) antibody that selectively binds to DLL4. Clinical results are undisclosed at the time of writing (Table 2).

Notch signaling affects lineage-specific differentiation and self-renewal of normal human breast stem cells[92]. Moreover, Notch-4 activity is elevated in breast CSC, and inhibiting Notch4 activity can reduce the breast CSC population, thereby suppressing tumor initiation[93]. Cleavage of the Notch receptor by gamma-secretase is required to release the Notch intracellular domain, which then translocates to the nucleus, turning on genes involved in cell differentiation and proliferation. Thus, gamma secretase inhibition results in a loss of Notch function in cells. In breast cancer xenografts, the gamma-secretase inhibitor MK-0752 reduced CSC and enhanced the efficacy of docetaxel[94]. Subsequently, 30 patients with locally advanced or metastatic breast cancer were treated with escalating doses of oral MK-0752[95] plus docetaxel every 3 wk. All patients could be evaluated for toxicity. Overall, 12/30 patients experienced G3 adverse reactions including G3 diarrhea (2 patients) and G3 liver function tests elevation at the selected dose level. Further, 24 patients could be evaluated for efficacy. In 11/24 patients, partial remissions were recorded, and in 9/24 patients, stable diseases were recorded. Since docetaxel has an established activity against breast cancer[96], the response rate should be interpreted with caution. Tumor biopsies were optional for participants in the study, and were performed more frequently in patients with locally advanced disease for whom surgery following initial medical treatment was planned. Serial biopsies were taken from 6 patients, at baseline, after cycle 1 and 3 and at progression or completion of treatment. All patients but one were treated at the selected dose level. Four patients experienced Partial Response, and 2 patients achieved stable disease. CSC were evaluated by flow cytometry examining both ALDH+ and CD44+/CD24- cell populations and by mammosphere forming efficiency. CD44+/CD24- cells decreased from baseline to surgery in 3/5 patients, while ALDH+ cells decreased from baseline to surgery in 3/6 patients. Mammosphere forming efficiency decreased over the course of treatment[94]. Despite these results, later phase clinical trials with MK-0752 are not listed on http://clinicaltrials.gov, neither for breast cancer nor for any other tumor type.

Wnt signaling pathway

The Wnt signaling pathway is another developmental pathway involved in multiple biological processes including embryogenesis, development, cell proliferation, survival and differentiation[97]. The Wnt/β-catenin signaling pathway is the best characterized one of the Wnt pathways[74]. Wnt/β-catenin signaling is activated when secreted glycoprotein, Wnt ligand, binds to the cell membrane Frizzled receptors and LRP5/6 co-receptors, which results in triggering of target genes[97]. Alteration in Wnt/β-catenin signaling has been identified in various malignancies, such as leukemia, colon, breast and cutaneous carcinoma. Wnt signaling in many human tumors arises from mutations in pathway components, resulting in aberrantly high levels of activation. For example, defective mutations in the adenomatous polyposis coli gene, a key downstream regulator of Wnt signaling, result in the inappropriate stabilization of β-catenin, thus activating the Wnt cascade and inducing epithelial cell transformation in patients with familial adenomatous polyposis[98,99]. The Wnt signaling pathway is associated with the maintenance of CSC stem cell properties, but it also regulates important stem cell niches within the body, including normal hematopoietic stem cells and adult stem cells within various tissues.

OMP-18R5 (vantictumab) (Table 2), a fully human IgG2 monoclonal antibody targeting the Frizzled receptors 1, 2, 5, 7 and 8, displays strong anti-tumor activity in patient-derived xenografts in association with standard of care chemotherapy[100]. It also induces cancer cell differentiation. A phase Ib study of the single agent OMP-18R5 in 25 patients demonstrated pharmacodynamic activity on hair follicles and bone turnover. Prolonged stabilization of disease was recorded in 3 patients with neuroendocrine tumors. Analysis of pre- and post-treatment tumor biopsies showed inhibition of Wnt and CSC genes, as well as upregulation of differentiation genes[101].

OMP-54F28 is a fusion protein based on a truncated form of the Frizzled8 receptor, or Fzd8, and the Fc fragment of human immunoglobulin IgG1. It is designed to bind Wnt ligands thus blocking Wnt signaling. In patient-derived xenograft models, OMP-54F28 acts synergetically with chemotherapy in a broad spectrum of malignancies. This fusion protein was tested as a single agent in a phase I study in patients with advanced solid tumors[102]. Intravenous injections every 3 wk did not result in any grade ≥ 3 adverse reactions at doses up to 20 mg/kg, double the target efficacious dose established by animal data and pharmacokinetic modeling. Most common (> 20% of patients) grade 1-2 adverse reactions were dysgeusia, decreased appetite, fatigue, muscle spasms, nausea and vomiting. No objective responses were recorded in 25 patients treated in 7 dose escalation cohorts, whereas several patients experienced prolonged stabilization of the disease. Pharmacodynamic modulation of Wnt pathway genes was shown in hair follicles. Five patients experienced doubling of the bone turnover marker β-C-terminal telopeptide, consistent with Wnt pathway inhibition in bone. This adverse reaction was easily managed with a single dose of zoledronic acid. Phase Ib studies of OMP-54F28 in combination with gemcitabine and nab-paclitaxel (pancreatic cancer), carboplatin and paclitaxel (ovarian cancer) and sorafenib (hepatocellular carcinoma) are currently in progress (Table 2).

CXCR1/2-CXCL8 signaling pathway

CXCL8 [formerly interleukin (IL-8)] is a small soluble protein belonging to the chemokine family[103]. It was originally identified as a potent neutrophil activator and chemotactic factor mainly secreted by activated monocytes and macrophages[104,105]. The biological effects of CXCL8 are mediated by two class A, rhodopsin-like guanine-protein-coupled receptors: CXC chemokine receptor 1 (CXCR1) (IL-8RA) and CXCR2 (IL-8RB)[106,107]. CXCL8 is upregulated in a wide variety of solid cancers, such as prostate, gastric, bladder, ovarian, lung cancer and melanoma. It has also been reported to contribute to multiple hallmarks of cancer, such as increased proliferation, angiogenesis, invasion, and metastases[108,109]. CXCL8 is overexpressed in breast cancer, compared with normal breast tissue, and although there is substantial evidence that CXCL8 may promote breast cancer initiation and progression, more recent evidence indicates that this cytokine is a key regulator of CSC activity[110]. A search for actionable molecules on ALDH+ breast cancer CSC identified CXCR1 as a target almost exclusively expressed by CSC as compared with bulk tumor cells[47]. It was shown in vitro that breast cancer CSC proliferate in response to exogenous CXCL8 and that addition of a small molecular weight antagonist of CXCR1/2 (reparixin, formerly repertaxin)[111] or a blocking anti-CXCR1 (but not anti-CXCR2) monoclonal antibody depleted CSC in vitro[56]. Interestingly, a FAS-FASL mediated bystander effect killed the vast majority of bulk tumor cells in vitro, suggesting the possibility of synergistic effects with chemotherapy[56]. In breast cancer patient-derived xenografts or human breast cancer cell lines, the combination of docetaxel and reparixin was more effective than chemotherapy alone in reducing tumor size[56]. As expected, administration of the single agent reparixin did not result in tumor shrinkage over a 4 wk treatment period. However, tumors recovered from mice that had been treated with reparixin alone or in combination with chemotherapy displayed a far lower abundance of CSC than tumors recovered from mice receiving chemotherapy alone[56]. In addition, reparixin reduced metastasis formation in mice following injection of luciferase-transfected human breast cancer cells into the bloodstream[56]. These results were integrated into a model in which following administration of chemotherapy, dying bulk tumor cells release CXCL8 and FASL. However, CXCR1+ CSC are sheltered from apoptotic signals from FASL unless CXCR1 is blocked by reparixin. Reparixin is a clinical grade molecule already used in patients with non-oncological conditions[112]. Therefore, clinical trials of oral reparixin were started in combination with weekly paclitaxel in HER2-negative pretreated metastatic breast cancer (NCT02001974)[113], and as a single agent in a window of opportunity trial in HER2-negative operable breast cancer (NCT01861054)[114]. Indirect support for this model comes from two lines of evidence. First, Bolha et al[55] reported release of CXCL8 from tumor cells exposed to taxane in vitro. Also, they observed a marked and dose-dependent increase in mammosphere forming efficiency in TNBC tumor cells recovered from immunocompromised mice treated with 2 doses of paclitaxel[55]. Second, Singh and colleagues[115] demonstrated measurable IL-8 levels in pleural effusions and ascites from breast cancer patients When tumor cells from pleural effusions and ascites were cultured in vitro, a direct correlation between IL-8 levels and CSC activity could be measured by mammosphere formation[115]. Surface CXCR1 was detected on the majority of mammosphere cells, and a CXCR1/2 inhibitor, SCH563705, blocked the effects of exogenous CXCL8 on mammosphere formation[115]. In partial contrast to findings by Ginestier pointing to a role for CXCR1 but not CXCR2 in anti-CSC activity[56], CXCR2 inhibition was shown to lead to a significant growth inhibition of basal like breast cancer cell lines in vitro[116], further highlighting the potential of CXCR1/2 inhibition. The activity was far less pronounced on non-basal like breast cancer cell lines. Also, in the report by Ginestier, most of the cell lines and patient-derived xenografts were triple negative breast cancers[56]. Although basal like and triple negative breast cancers do not fully overlap[44], these findings suggest that basal like and/or triple negative breast cancer could be most sensitive to CXCR1/2 inhibition, possibly due to an enrichment in CSC in these breast cancer types[42-44].

A possible role for CXCL8 on CSC has been recently highlighted also in pancreatic cancer[53]. The authors found a positive correlation between CXCR1 and both CD44 and CD133 expression in human pancreatic cancer samples. Furthermore, addition of exogenous CXCL8 in vitro increased sphere formation, CSC population, and cell invasion of pancreatic cancer cells, all these effects being reversible upon addition of a CXCR1-blocking monoclonal antibody[53].

CXCR1/2 blockade may lead to anti-CSC effects also by an indirect mechanism. In fact, mesenchymal stem cells, a stromal cell type secreting factors that sustain CSC, are often recruited to tumors by tumor cell-derived CXCL8[117]. Thus, inhibition of CXCL8 activity and its homing signal may prevent localization of mesenchymal stem cells to the tumor stroma, hindering the development of a supportive CSC niche[15].

Finally, considering the multiple roles played by CXCL8 in cancer (reviewed in[118]), it should be borne in mind that inhibition of CXCL8 biological activity may lead to anti-tumor activity also by other mechanisms not directly related to CSC, such as inhibition of angiogenesis and modification of the leukocyte infiltration into tumors.

IL-6/Janus kinase 2/Signal transducer and activator of transcription 3

The IL-6/Janus kinase 2 (JAK2)/Signal transducer and activator of transcription 3 (Stat3) pathway was found to be preferentially activated in CD44+/CD24- breast CSC[116]. This opens the possibility of targeting CSC by compounds interfering with any of the 3 pathway components. Approved anti-IL-6 compounds (tocilizumab, anti-IL-6R mAb) and JAK inhibitors (i.e., tofacitinib) are available for the treatment of rheumatoid arthritis, thus making this pathway a readily available clinical target. Furthermore, it has been shown in preclinical studies that breast cancer cell resistance to trastuzumab, an approved monoclonal antibody for treatment of HER2+ breast cancer, is mediated by an IL-6 feedback loop, leading to an expansion of the CSC pool[119]. Indirect support for a role played by JAK2 comes also from preclinical studies using chloroquine (another approved drug for treating malaria), which suppressed CD44+/CD24- breast CSC via inhibition of the JAK2-Stat3 pathway[57].

HER2/AKT

HER2 has been reported to be an intrinsic regulator of breast cancer CSC[120,121]. This regulation occurs through activation of the Wnt/β-catenin pathway via Akt mediated phosphorylation of GSK3B and nuclear translocation of β-catenin[122]. In patients with HER2-positive breast cancer undergoing neoadjuvant treatment, lapatinib administration led to a decrease, although not a statistically significant one, in both the proportion of CD44+/CD24- CSC and mammosphere forming efficiency[58]. However, HER2 regulation of CSC possibly extends to HER2-negative breast cancer (reviewed in[123], as suggested by the clinical benefit observed in patients with HER2-negative breast cancer receiving adjuvant anti-HER2 treatment[124]. It has been shown that in HER2-negative breast cancer, CSC selectively express, and are regulated by, HER2[125]. This is in line with the observation that in vitro, lapatinib inhibits CXCL8-stimulated mammosphere formation from both HER2-positive and HER2-negative patient breast cancer samples, highlighting a key role of HER2 in regulating CXCR1/2 mediated activation of breast cancer CSC[115]. This data was put into context in a model in which CSC activity is increased following CXCL8 binding to CXCR1/2 via both HER2-independent and HER2-dependent (through CXCR1/2 mediated transactivation) mechanisms[115]. From a clinical standpoint, this data suggests that combination therapy with anti-HER2 and anti-CXCR1/2 compounds may affect breast cancer CSC to a greater extent than either compound alone[115,126].

REPURPOSED DRUGS

A number of approved drugs that have been used for many years in non-oncological indications have been shown to have anti-tumor activity in vitro and/or in vivo. The obvious advantage of such drugs is the extensive knowledge of their safety profile (although not in association with standard of care chemotherapy), which may open the possibility for single agent window of opportunity trials in a preoperative setting for selected cancers, or possibly neoadjuvant trials if overlapping toxicities are not to be expected[127]. In other instances, high throughput screening programs have identified known compounds as potential CSC targeting agents[128,129]. Several such drugs are being tested as anti-CSC.

Chloroquine is an antimalarial drug. It is capable of inhibiting autophagy, which is a mechanism exploited by cancer cells to cope with stress[130]. In particular, autophagy has emerged as a mechanism by which CSC maintain their capability of self-renewal[131]. In silico gene expression analysis of the CD44+/CD24- treatment resistant breast cancer cell population identified chloroquine as a candidate anti-CSC compound[57]. In vitro chloroquine reduced, in a dose-dependent manner, mammosphere forming efficiency in TNBC cell lines as well as CD44+/CD24- cells when used alone or in combination with paclitaxel[57]. In vivo, growth of orthotopically implanted tumors as well as lung metastases were reduced, and so were CSC when chloroquine was administered in combination with paclitaxel[57]. A clinical trial (NCT01446016) is currently in progress, in which chloroquine is administered in combination with a taxane or taxane-like drug (i.e., nab-paclitaxel and ixabepilone) to women with advanced or metastatic breast cancer, who have not responded to anthracycline therapy. An interim report showed that in two out of three patients, from whom pre- and post-treatment biopsies were taken, a clear reduction in CD44+/CD24- cells was recorded[57]. Chloroquine has been investigated as a CSC targeting agent not only in breast cancer but also in CML[132].

A significant number of studies has been performed with the anti-diabetic drug metformin[133]. Its activity against CSC has been described in a number of preclinical reports[134,135], whereas the issue of its clinical activity against CSC has remained unexplored despite a number of window of opportunity studies in patients with operable breast cancer that established safety of the drug in this setting[136].

Itraconazole and arsenic trioxide, two drugs approved for fungal infections and acute promyelocytic leukemia, respectively, have been shown to be Hedgehog pathway inhibitors[137,138]. In an open label phase 2 clinical trial in patients with basal cell carcinoma, a 24% decrease in tumor area, a 65% reduction in Hedgehog pathway activity and a 45% reduction in the proliferative marker Ki-67 were observed following treatment with itraconazole in comparison with untreated patients[139]. The potential for all-trans retinoic acid as an anti-CSC agent has been recently and thoroughly reviewed[140].

Salinomycin, which is used in chicken fodder as a coccidiostat, has been shown to reduce the proportion of CSC by > 100-fold relative to paclitaxel, a commonly used breast cancer chemotherapeutic drug[128]. Treatment of mice with salinomycin inhibits mammary tumor growth in vivo and induces increased epithelial differentiation of tumor cells[128]. In addition, global gene expression analyses show that salinomycin treatment results in loss of expression of breast CSC genes previously identified by analyses of breast tissues isolated from patients[128]. However, no clinical trials testing salinomycin are registered on http://clinicaltrials.gov.

Thioridazine, an antipsychotic drug, selectively targets leukemic stem cells while sparing normal hematopoietic stem cells[129]. The drug antagonizes dopamine receptors that are expressed on CSC[129]. It is currently being tested in a phase I trial in combination with cytarabine in patients with acute myeloid leukemia (NCT02096289).

ISSUES IN DESIGNING CLINICAL TRIALS FOR CSC-TARGETING AGENTS

Early drug development in cancer usually requires signs of anti-tumor activity as measured by shrinkage of metastatic lesions. Once this is demonstrated, a novel agent is then moved to the adjuvant setting, i.e., after removal of the primary tumor, to reduce recurrence risk. Based on the CSC model, and considering that CSC represent only a tiny percentage of the total tumor cells (i.e., < 5%) of any given tumor, single agent CSC-specific agents would not be expected to display antitumor activity measurable by RECIST criteria[141]. As a consequence, in metastatic patients such agents would not be expected to affect tumor growth and tumor-related symptoms in the short term. Thus, in this setting, CSC-specific agents likely need be tested in combination with standard of care chemotherapy that will address the proliferating, non-CSC portion of the tumor. When a CSC-targeting agent is used in combination with standard of care chemotherapy, the objective response rate is still an obvious endpoint, but its relevance is mostly to ensure that the CSC-targeting agent does not hinder chemotherapy activity. In fact, it is unlikely that a specific CSC-targeting agent augments chemotherapy activity as measured by the objective response rate unless indirectly mediated through a bystander effect on bulk tumor cells[56].

The same consideration applies to neoadjuvant chemotherapy, the purpose of which is to downsize the primary tumor and to eliminate micrometastases. Also, in this setting, it is not intuitive that a CSC-specific agent can significantly increase the rate of pCR as compared to chemotherapy alone, unless a bystander effect is postulated. In this specific setting, one may hypothesize that in a randomized clinical trial, the rate of pCR as well as complete and partial clinical responses between the two treatment groups could be very similar, but disease-free survival may differ, favoring the regimen including a CSC-targeting agent. Partial support for this hypothesis comes from the study by Alamgeer et al[61], in which patients who experienced less than pCR but had eliminated ALDH+ CSC in their primary tumor following neoadjuvant chemotherapy had the same disease-free survival as patients achieving a pCR.

Therefore, the objective response rate per se is not necessarily indicative of anti-CSC activity, whereas the duration of response would be more informative. These considerations are relevant for the identification of effective compounds for adjuvant treatment, in which activity on CSC is expected to be most relevant, according to the CSC paradigm[123]. Evaluation of pre- and post-treatment biopsies is conceivably the most compelling evidence for anti-CSC activity. However, post-treatment biopsy can be difficult to obtain from metastatic patients[94]. Other challenges also may hinder the value of repeated biopsies even in the neoadjuvant setting, in which tumor tissue is readily available. In breast cancer, the most widely used CSC markers are ALDH+ and CD44+/CD24-. However, it has been shown that cells displaying either phenotype are largely not overlapping, with each marker(s) identifying a distinct CSC population[41,48,49]. Furthermore, breast cancer CSC can transition from one phenotype to the other. Little is known about the role, and the therapeutic relevance, of each CSC population in primary breast cancer. However, recent evidence shows that ALDH+ cells are proliferative, epithelial cell-like CSC, whereas CD44+/CD24- CSC are quiescent, mesenchymal-like cells poised to invade and metastasize[49]. In keeping with their different putative role in cancer, ALDH+ and CD44+/CD24- cells tend to be localized in different areas of the tumor, i.e., center and edges, respectively[49]. The location of these CSC populations in anatomically distinct areas of a tumor, coupled with their plasticity, poses theoretical challenges to comparative measurement of CSC in serial biopsies taken before, during and after treatment.

In drug development, in order to treat patients with potentially curable diseases with a novel agent, a robust safety database from the metastatic setting or from other indications for repurposed drugs must be available. Thus, most novel agents designed to target CSC will be tested first in patients with metastatic disease due to safety reasons. One may argue that the metastatic setting is less than ideal to test CSC-targeting agents, since they are conceivably best suited to prevent or reduce metastatic spread and thus should be applied to the (neo)adjuvant setting. In order to circumvent these limitations, surrogate clinical endpoints have been devised to evaluate the potential of a novel agent to reduce the metastatic spread of tumors[142]. This concept fits into the CSC model and can be applied to the development of a CSC-targeting agent which, as a novel chemical entity or biologic, cannot be tested in settings of earlier disease stages due to safety concerns. Time to new metastasis (TTM) is an exploratory endpoint that measures the time elapsing from study entry or randomization to detection of metastatic lesions in a new organ system as compared to baseline[143]. Examples of application of this endpoint can be found in lung cancer[144,145]. In a less stringent fashion, this endpoint can be measured also until appearance of new lesion(s) in an organ already metastatic at baseline. In breast cancer, considering the proportion of patients developing either new metastatic sites or new lesions at their first tumor progression[146], this endpoint can certainly be read with the possibility of demonstrating the potential for a novel agent to reduce metastatic spread. It should be noted that TTM has also immediate clinical relevance since the development of new metastases in the brain, bones or viscera can lead to significant morbidity. The data with denosumab in the prevention of bone metastases in patients with castration-resistant, non-metastatic prostate cancer[147], as well as the reduction of skeletal-related events in patients with metastatic breast cancer[148], is reminiscent of this endpoint. However, TTM is a challenging endpoint to measure, as in several metastatic tumors, patients with progression will be put on another treatment so that TTM can be read only until RECIST 1.1 progression[143]. The most thorough way to assess TTM would be to design trials allowing treatment beyond progression[143]. One possible way to measure anti-CSC activity circumventing the need for serial biopsies would be to analyze cancer cells and/or tumor DNA in patients’ bloodstream. The prognostic role of circulating tumor cells (CTCs) has been established since 2004[149,150]. However, the relationship between the number of CTC and tumor-localized or circulating CSC has not been established yet. The potential, but also the current pitfalls, of CTC and circulating DNA have been recently reviewed elsewhere[151,152].

Targeted anti-cancer therapies are best developed in conjunction with biomarkers (e.g., crizotinib, vemurafenib) that can measure treatment efficacy and/or can identify patients with a higher chance to benefit from the treatment. As discussed earlier, not all tumors display a sizeable CSC population, although this finding may be related to the sensitivity of the assay used and the use of a single marker. The ideal biomarker for pharmacodynamic evaluation of cancer stem cell therapies would be able to accurately measure the proportion of stem cells within the tumor, so as to select patients with a high proportion of stem cells for treatment, to identify presence of the target on CSC for targeted therapies, and to determine on pre- and post-therapy specimens (i.e., tumor or blood) whether the proportion of stem cells within the tumor decreased under the therapy. Unfortunately, the tiny percentage of CSC in a tumor mass makes it difficult to develop a companion diagnostic for any CSC targeting agent with a molecular target.

CONCLUSION

The existence of multiple subpopulations of cells within a tumor with distinct tumor initiating capacity has been clearly demonstrated[14,15]. The clinical relevance of CSC is being highlighted by both retrospective analyses and prospective studies, challenging clinical paradigms[61]. There is still controversy about the most suitable markers to identify CSC in order to understand their role and therapeutic relevance in different stages of human cancers, as well as about monitoring effects of CSC targeting agents. There is a requirement to identify cell surface marker and gene activity profiles that can be readily and reliably used to identify CSC in human tumors. However, as these genes frequently operate in other cell types, they cannot be called “stemness” genes. In order to identify true signatures and pure populations, microarray and genome-wide techniques can be applied to detect trends in genetic and epigenetic models for cancer stem cells. This is especially true for cells expected to be rare, such as cancer stem cells, the expression signature of which would be saturated by the majority of non-stem cells in a whole tumor sample. Even after a cancer stem cell signature from a particular type of tumor is identified, one cannot assume that a given signature is useful for identifying cancer stem cells in a different tumor type, unless it is validated by a functional assay. The use of gene inactivation to eliminate “stemness” or gene activation to engender “stemness” would be required to functionally link any marker to stem cell identity. This can be applied to clinical trials as exemplified by the study that identified RPL39 and MFL2 as potential targets in CSC[153].

A number of novel or repurposed agents interfering with pathways considered relevant for CSC are being evaluated in patients with advanced tumors following demonstration of activity in patient-derived xenografts. A recommended phase 2 dose has been identified for most of such agents either alone or in combination with chemotherapy. For some agents, pharmacodynamic demonstration of target pathway modulation of non-cancer targets (e.g., hair follicles) has been obtained, and some long term responders or at least non-progressors have been described. Formal demonstration of an anti-CSC effect is still lacking in most cases, due to a number of factors including the challenge to obtain serial biopsies in metastatic patients, and the interference of several anti-CSC agents with multiple pathways makes it difficult to disentangle the relative contribution of the anti-CSC vs non-anti-CSC activity. Clinical surrogate endpoints may prove useful in suggesting an anti-CSC activity, such as reduction of metastatic spread measured as time to new metastasis[143]. However, in order to seek marketing authorization, a CSC-targeting agent should demonstrate an advantage at least in terms of Progression-Free Survival and Disease-Free Survival as compared with standard of care in the metastatic and (neo)adjuvant setting, respectively. In this respect, the goals recently put forward by the American Society of Clinical Oncology Cancer Research Committee[154] may be within reach if the CSC model holds true for a given malignancy. Achievement of these goals raises the issue of patient selection, a rather unexplored issue thus far, since trials to date have been conducted on unselected patient populations. First, it is not known whether the CSC paradigm is universal to all human cancers[14]. Second, it has been clearly established that administration of chemotherapy in the adjuvant setting (i.e., following surgery of the primary tumor) increases 30-year disease-free survival as compared with surgery alone in node-positive breast cancer patients[155]. This suggests that a fraction of patients does not seem to require a CSC targeting agent, whereas the opposite is entirely possible in another set of patients or a different breast cancer subtype. Third, aberrant activation of any of the pathways discussed here due to mutations, as seen in a proportion of tumors, may represent one selection step. Lastly, it is possible that CSC may eventually show resistance to CSC targeting agents. At least two drug resistance pathways have been identified for the Hedgehog inhibitor vismodegib in patients[156,157].

In conclusion, given the extraordinary complexity of CSC clinical targeting, a bench to bedside to bench circle[94] is the most sensible approach to develop anti-CSC agents.

Footnotes

P- Reviewer: de la Serna IL, Ho I, Liu L, Safa AR S- Editor: Song XX L- Editor: A E- Editor: Liu SQ

References
1.  D’Angelo RC, Wicha MS. Stem cells in normal development and cancer. Prog Mol Biol Transl Sci. 2010;95:113-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
2.  Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, Wahl GM. Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 2006;66:9339-9344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2176]  [Cited by in F6Publishing: 2134]  [Article Influence: 118.6]  [Reference Citation Analysis (0)]
3.  Alison MR, Lim SM, Nicholson LJ. Cancer stem cells: problems for therapy? J Pathol. 2011;223:147-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 219]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
4.  Passegué E, Jamieson CH, Ailles LE, Weissman IL. Normal and leukemic hematopoiesis: are leukemias a stem cell disorder or a reacquisition of stem cell characteristics? Proc Natl Acad Sci USA. 2003;100 Suppl 1:11842-11849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 456]  [Cited by in F6Publishing: 434]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
5.  Cobaleda C, Gutiérrez-Cianca N, Pérez-Losada J, Flores T, García-Sanz R, González M, Sánchez-García I. A primitive hematopoietic cell is the target for the leukemic transformation in human philadelphia-positive acute lymphoblastic leukemia. Blood. 2000;95:1007-1013.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  O’Connor ML, Xiang D, Shigdar S, Macdonald J, Li Y, Wang T, Pu C, Wang Z, Qiao L, Duan W. Cancer stem cells: A contentious hypothesis now moving forward. Cancer Lett. 2014;344:180-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 176]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
7.  Wang JC, Dick JE. Cancer stem cells: lessons from leukemia. Trends Cell Biol. 2005;15:494-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 433]  [Cited by in F6Publishing: 463]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
8.  Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730-737.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100:3983-3988.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7830]  [Cited by in F6Publishing: 7472]  [Article Influence: 355.8]  [Reference Citation Analysis (0)]
10.  O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445:106-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2977]  [Cited by in F6Publishing: 2949]  [Article Influence: 163.8]  [Reference Citation Analysis (0)]
11.  Frank NY, Schatton T, Frank MH. The therapeutic promise of the cancer stem cell concept. J Clin Invest. 2010;120:41-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 452]  [Cited by in F6Publishing: 488]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
12.  Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593-598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1362]  [Cited by in F6Publishing: 1436]  [Article Influence: 95.7]  [Reference Citation Analysis (0)]
13.  De Palma M, Venneri MA, Galli R, Sergi Sergi L, Politi LS, Sampaolesi M, Naldini L. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell. 2005;8:211-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1009]  [Cited by in F6Publishing: 957]  [Article Influence: 50.4]  [Reference Citation Analysis (0)]
14.  Dick JE. Stem cell concepts renew cancer research. Blood. 2008;112:4793-4807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 736]  [Cited by in F6Publishing: 717]  [Article Influence: 47.8]  [Reference Citation Analysis (0)]
15.  Pattabiraman DR, Weinberg RA. Tackling the cancer stem cells - what challenges do they pose? Nat Rev Drug Discov. 2014;13:497-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 660]  [Cited by in F6Publishing: 737]  [Article Influence: 73.7]  [Reference Citation Analysis (0)]
16.  Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu Rev Pathol. 2006;1:119-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 726]  [Cited by in F6Publishing: 775]  [Article Influence: 43.1]  [Reference Citation Analysis (0)]
17.  Clarke MF, Fuller M. Stem cells and cancer: two faces of eve. Cell. 2006;124:1111-1115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 633]  [Cited by in F6Publishing: 626]  [Article Influence: 34.8]  [Reference Citation Analysis (0)]
18.  Feng Y, Wen J, Mike P, Choi DS, Eshoa C, Shi ZZ, Zu Y, Chang CC. Bone marrow stromal cells from myeloma patients support the growth of myeloma stem cells. Stem Cells Dev. 2010;19:1289-1296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
19.  Al-Hajj M, Becker MW, Wicha M, Weissman I, Clarke MF. Therapeutic implications of cancer stem cells. Curr Opin Genet Dev. 2004;14:43-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 433]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
20.  Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821-5828.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Corbeil D, Röper K, Hellwig A, Tavian M, Miraglia S, Watt SM, Simmons PJ, Peault B, Buck DW, Huttner WB. The human AC133 hematopoietic stem cell antigen is also expressed in epithelial cells and targeted to plasma membrane protrusions. J Biol Chem. 2000;275:5512-5520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 325]  [Cited by in F6Publishing: 321]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
22.  Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432:396-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5422]  [Cited by in F6Publishing: 5376]  [Article Influence: 268.8]  [Reference Citation Analysis (0)]
23.  Keysar SB, Jimeno A. More than markers: biological significance of cancer stem cell-defining molecules. Mol Cancer Ther. 2010;9:2450-2457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 163]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
24.  Zhang Q, Shi S, Yen Y, Brown J, Ta JQ, Le AD. A subpopulation of CD133(+) cancer stem-like cells characterized in human oral squamous cell carcinoma confer resistance to chemotherapy. Cancer Lett. 2010;289:151-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 182]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
25.  Bertolini G, Roz L, Perego P, Tortoreto M, Fontanella E, Gatti L, Pratesi G, Fabbri A, Andriani F, Tinelli S. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc Natl Acad Sci USA. 2009;106:16281-16286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 562]  [Cited by in F6Publishing: 617]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
26.  Moreb JS. Aldehyde dehydrogenase as a marker for stem cells. Curr Stem Cell Res Ther. 2008;3:237-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 190]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
27.  Storms RW, Trujillo AP, Springer JB, Shah L, Colvin OM, Ludeman SM, Smith C. Isolation of primitive human hematopoietic progenitors on the basis of aldehyde dehydrogenase activity. Proc Natl Acad Sci USA. 1999;96:9118-9123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 398]  [Cited by in F6Publishing: 388]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
28.  Silva IA, Bai S, McLean K, Yang K, Griffith K, Thomas D, Ginestier C, Johnston C, Kueck A, Reynolds RK. Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 2011;71:3991-4001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 360]  [Cited by in F6Publishing: 387]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
29.  Kristiansen G, Sammar M, Altevogt P. Tumour biological aspects of CD24, a mucin-like adhesion molecule. J Mol Histol. 2004;35:255-262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 231]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
30.  Sano A, Kato H, Sakurai S, Sakai M, Tanaka N, Inose T, Saito K, Sohda M, Nakajima M, Nakajima T. CD24 expression is a novel prognostic factor in esophageal squamous cell carcinoma. Ann Surg Oncol. 2009;16:506-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 54]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
31.  Aigner S, Sthoeger ZM, Fogel M, Weber E, Zarn J, Ruppert M, Zeller Y, Vestweber D, Stahel R, Sammar M. CD24, a mucin-type glycoprotein, is a ligand for P-selectin on human tumor cells. Blood. 1997;89:3385-3395.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Croker AK, Goodale D, Chu J, Postenka C, Hedley BD, Hess DA, Allan AL. High aldehyde dehydrogenase and expression of cancer stem cell markers selects for breast cancer cells with enhanced malignant and metastatic ability. J Cell Mol Med. 2009;13:2236-2252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 350]  [Cited by in F6Publishing: 365]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
33.  Klarmann GJ, Hurt EM, Mathews LA, Zhang X, Duhagon MA, Mistree T, Thomas SB, Farrar WL. Invasive prostate cancer cells are tumor initiating cells that have a stem cell-like genomic signature. Clin Exp Metastasis. 2009;26:433-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 141]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
34.  Hurt EM, Kawasaki BT, Klarmann GJ, Thomas SB, Farrar WL. CD44+ CD24(-) prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis. Br J Cancer. 2008;98:756-765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 316]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
35.  Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67:1030-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2377]  [Cited by in F6Publishing: 2352]  [Article Influence: 138.4]  [Reference Citation Analysis (0)]
36.  Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3316]  [Cited by in F6Publishing: 3222]  [Article Influence: 107.4]  [Reference Citation Analysis (0)]
37.  Zhou L, Jiang Y, Yan T, Di G, Shen Z, Shao Z, Lu J. The prognostic role of cancer stem cells in breast cancer: a meta-analysis of published literatures. Breast Cancer Res Treat. 2010;122:795-801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 81]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
38.  Liu Y, Lv DL, Duan JJ, Xu SL, Zhang JF, Yang XJ, Zhang X, Cui YH, Bian XW, Yu SC. ALDH1A1 expression correlates with clinicopathologic features and poor prognosis of breast cancer patients: a systematic review and meta-analysis. BMC Cancer. 2014;14:444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 70]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
39.  Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA. Molecular portraits of human breast tumours. Nature. 2000;406:747-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10665]  [Cited by in F6Publishing: 10416]  [Article Influence: 434.0]  [Reference Citation Analysis (0)]
40.  Charafe-Jauffret E, Ginestier C, Iovino F, Tarpin C, Diebel M, Esterni B, Houvenaeghel G, Extra JM, Bertucci F, Jacquemier J. Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res. 2010;16:45-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 514]  [Cited by in F6Publishing: 535]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
41.  Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu S. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2860]  [Cited by in F6Publishing: 2939]  [Article Influence: 183.7]  [Reference Citation Analysis (0)]
42.  Hennessy BT, Gonzalez-Angulo AM, Stemke-Hale K, Gilcrease MZ, Krishnamurthy S, Lee JS, Fridlyand J, Sahin A, Agarwal R, Joy C. Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res. 2009;69:4116-4124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 624]  [Cited by in F6Publishing: 632]  [Article Influence: 42.1]  [Reference Citation Analysis (0)]
43.  Idowu MO, Kmieciak M, Dumur C, Burton RS, Grimes MM, Powers CN, Manjili MH. CD44(+)/CD24(-/low) cancer stem/progenitor cells are more abundant in triple-negative invasive breast carcinoma phenotype and are associated with poor outcome. Hum Pathol. 2012;43:364-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
44.  Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363:1938-1948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2585]  [Cited by in F6Publishing: 2743]  [Article Influence: 195.9]  [Reference Citation Analysis (0)]
45.  Mayer IA, Abramson VG, Lehmann BD, Pietenpol JA. New strategies for triple-negative breast cancer--deciphering the heterogeneity. Clin Cancer Res. 2014;20:782-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 213]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
46.  Perou CM. Molecular stratification of triple-negative breast cancers. Oncologist. 2011;16 Suppl 1:61-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 254]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
47.  Charafe-Jauffret E, Ginestier C, Iovino F, Wicinski J, Cervera N, Finetti P, Hur MH, Diebel ME, Monville F, Dutcher J. Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer Res. 2009;69:1302-1313.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Liu Y, Nenutil R, Appleyard MV, Murray K, Boylan M, Thompson AM, Coates PJ. Lack of correlation of stem cell markers in breast cancer stem cells. Br J Cancer. 2014;110:2063-2071.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
49.  Liu S, Cong Y, Wang D, Sun Y, Deng L, Liu Y, Martin-Trevino R, Shang L, McDermott SP, Landis MD. Breast cancer stem cells transition between epithelial and mesenchymal states reflective of their normal counterparts. Stem Cell Reports. 2014;2:78-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 649]  [Cited by in F6Publishing: 726]  [Article Influence: 66.0]  [Reference Citation Analysis (0)]
50.  Liu S, Liu C, Min X, Ji Y, Wang N, Liu D, Cai J, Li K. Prognostic value of cancer stem cell marker aldehyde dehydrogenase in ovarian cancer: a meta-analysis. PLoS One. 2013;8:e81050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
51.  Le Magnen C, Bubendorf L, Rentsch CA, Mengus C, Gsponer J, Zellweger T, Rieken M, Thalmann GN, Cecchini MG, Germann M. Characterization and clinical relevance of ALDHbright populations in prostate cancer. Clin Cancer Res. 2013;19:5361-5371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
52.  Li XS, Xu Q, Fu XY, Luo WS. ALDH1A1 overexpression is associated with the progression and prognosis in gastric cancer. BMC Cancer. 2014;14:705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
53.  Chen L, Fan J, Chen H, Meng Z, Chen Z, Wang P, Liu L. The IL-8/CXCR1 axis is associated with cancer stem cell-like properties and correlates with clinical prognosis in human pancreatic cancer cases. Sci Rep. 2014;4:5911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 119]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
54.  Ran D, Schubert M, Pietsch L, Taubert I, Wuchter P, Eckstein V, Bruckner T, Zoeller M, Ho AD. Aldehyde dehydrogenase activity among primary leukemia cells is associated with stem cell features and correlates with adverse clinical outcomes. Exp Hematol. 2009;37:1423-1434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 64]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
55.  Bhola NE, Balko JM, Dugger TC, Kuba MG, Sánchez V, Sanders M, Stanford J, Cook RS, Arteaga CL. TGF-β inhibition enhances chemotherapy action against triple-negative breast cancer. J Clin Invest. 2013;123:1348-1358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 415]  [Cited by in F6Publishing: 446]  [Article Influence: 40.5]  [Reference Citation Analysis (0)]
56.  Ginestier C, Liu S, Diebel ME, Korkaya H, Luo M, Brown M, Wicinski J, Cabaud O, Charafe-Jauffret E, Birnbaum D. CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts. J Clin Invest. 2010;120:485-497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 564]  [Cited by in F6Publishing: 567]  [Article Influence: 40.5]  [Reference Citation Analysis (0)]
57.  Choi DS, Blanco E, Kim YS, Rodriguez AA, Zhao H, Huang TH, Chen CL, Jin G, Landis MD, Burey LA. Chloroquine eliminates cancer stem cells through deregulation of Jak2 and DNMT1. Stem Cells. 2014;32:2309-2323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 85]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
58.  Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst. 2008;100:672-679.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1319]  [Cited by in F6Publishing: 1327]  [Article Influence: 82.9]  [Reference Citation Analysis (0)]
59.  Lee HE, Kim JH, Kim YJ, Choi SY, Kim SW, Kang E, Chung IY, Kim IA, Kim EJ, Choi Y. An increase in cancer stem cell population after primary systemic therapy is a poor prognostic factor in breast cancer. Br J Cancer. 2011;104:1730-1738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 136]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
60.  Tanei T, Morimoto K, Shimazu K, Kim SJ, Tanji Y, Taguchi T, Tamaki Y, Noguchi S. Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin Cancer Res. 2009;15:4234-4241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 406]  [Cited by in F6Publishing: 443]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
61.  Alamgeer M, Ganju V, Kumar B, Fox J, Hart S, White M, Harris M, Stuckey J, Prodanovic Z, Schneider-Kolsky ME. Changes in aldehyde dehydrogenase-1 expression during neoadjuvant chemotherapy predict outcome in locally advanced breast cancer. Breast Cancer Res. 2014;16:R44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
62.  Gong C, Yao H, Liu Q, Chen J, Shi J, Su F, Song E. Markers of tumor-initiating cells predict chemoresistance in breast cancer. PLoS One. 2010;5:e15630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
63.  Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A, Rimm DL, Wong H, Rodriguez A, Herschkowitz JI. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA. 2009;106:13820-13825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1015]  [Cited by in F6Publishing: 1047]  [Article Influence: 69.8]  [Reference Citation Analysis (0)]
64.  Akunuru S, James Zhai Q, Zheng Y. Non-small cell lung cancer stem/progenitor cells are enriched in multiple distinct phenotypic subpopulations and exhibit plasticity. Cell Death Dis. 2012;3:e352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 98]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
65.  Gonçalves A, Pierga JY, Brain E, Tarpin C, Cure H, Esterni B, Boyer-Chammard A, Bertucci F, Jalaguier A, Houvenaeghel G. Abstract ot2-3-05: Avastem: A phase II randomized trial evaluating anti-cancer stem cell activity of pre-operative bevacizumab and chemotherapy in breast cancer. Cancer Res. 2012;24:OT2-3-05.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Conley SJ, Gheordunescu E, Kakarala P, Newman B, Korkaya H, Heath AN, Clouthier SG, Wicha MS. Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia. Proc Natl Acad Sci USA. 2012;109:2784-2789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 472]  [Cited by in F6Publishing: 478]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
67.  Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, Shi Q, Cao Y, Lathia J, McLendon RE. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell. 2009;15:501-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 994]  [Cited by in F6Publishing: 972]  [Article Influence: 64.8]  [Reference Citation Analysis (0)]
68.  Chen X, Iliopoulos D, Zhang Q, Tang Q, Greenblatt MB, Hatziapostolou M, Lim E, Tam WL, Ni M, Chen Y. XBP1 promotes triple-negative breast cancer by controlling the HIF1α pathway. Nature. 2014;508:103-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 522]  [Cited by in F6Publishing: 592]  [Article Influence: 59.2]  [Reference Citation Analysis (0)]
69.  Jensen RL. Brain tumor hypoxia: tumorigenesis, angiogenesis, imaging, pseudoprogression, and as a therapeutic target. J Neurooncol. 2009;92:317-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 209]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
70.  Heddleston JM, Hitomi M, Venere M, Flavahan WA, Yang K, Kim Y, Minhas S, Rich JN, Hjelmeland AB. Glioma stem cell maintenance: the role of the microenvironment. Curr Pharm Des. 2011;17:2386-2401.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6844]  [Cited by in F6Publishing: 6706]  [Article Influence: 291.6]  [Reference Citation Analysis (0)]
72.  Molofsky AV, Pardal R, Morrison SJ. Diverse mechanisms regulate stem cell self-renewal. Curr Opin Cell Biol. 2004;16:700-707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 237]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
73.  Varjosalo M, Taipale J. Hedgehog: functions and mechanisms. Genes Dev. 2008;22:2454-2472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 890]  [Cited by in F6Publishing: 896]  [Article Influence: 56.0]  [Reference Citation Analysis (0)]
74.  Maugeri-Saccà M, Zeuner A, De Maria R. Therapeutic targeting of cancer stem cells. Front Oncol. 2011;1:10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
75.  Corbit KC, Aanstad P, Singla V, Norman AR, Stainier DY, Reiter JF. Vertebrate Smoothened functions at the primary cilium. Nature. 2005;437:1018-1021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1114]  [Cited by in F6Publishing: 1097]  [Article Influence: 57.7]  [Reference Citation Analysis (0)]
76.  Merchant AA, Matsui W. Targeting Hedgehog--a cancer stem cell pathway. Clin Cancer Res. 2010;16:3130-3140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 379]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
77.  Song Z, Yue W, Wei B, Wang N, Li T, Guan L, Shi S, Zeng Q, Pei X, Chen L. Sonic hedgehog pathway is essential for maintenance of cancer stem-like cells in human gastric cancer. PLoS One. 2011;6:e17687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 129]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
78.  Tang SN, Fu J, Nall D, Rodova M, Shankar S, Srivastava RK. Inhibition of sonic hedgehog pathway and pluripotency maintaining factors regulate human pancreatic cancer stem cell characteristics. Int J Cancer. 2012;131:30-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 158]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
79.  Chen YJ, Sims-Mourtada J, Izzo J, Chao KS. Targeting the hedgehog pathway to mitigate treatment resistance. Cell Cycle. 2007;6:1826-1830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
80.  Von Hoff DD, LoRusso PM, Rudin CM, Reddy JC, Yauch RL, Tibes R, Weiss GJ, Borad MJ, Hann CL, Brahmer JR. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med. 2009;361:1164-1172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 852]  [Cited by in F6Publishing: 805]  [Article Influence: 53.7]  [Reference Citation Analysis (0)]
81.  Sekulic A, Migden MR, Oro AE, Dirix L, Lewis KD, Hainsworth JD, Solomon JA, Yoo S, Arron ST, Friedlander PA. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N Engl J Med. 2012;366:2171-2179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 947]  [Cited by in F6Publishing: 950]  [Article Influence: 79.2]  [Reference Citation Analysis (0)]
82.  Tang JY, Mackay-Wiggan JM, Aszterbaum M, Yauch RL, Lindgren J, Chang K, Coppola C, Chanana AM, Marji J, Bickers DR. Inhibiting the hedgehog pathway in patients with the basal-cell nevus syndrome. N Engl J Med. 2012;366:2180-2188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 404]  [Cited by in F6Publishing: 379]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
83.  Amakye D, Jagani Z, Dorsch M. Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat Med. 2013;19:1410-1422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 405]  [Cited by in F6Publishing: 420]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
84.  Soltanian S, Matin MM. Cancer stem cells and cancer therapy. Tumour Biol. 2011;32:425-440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 108]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
85.  Wang Z, Li Y, Banerjee S, Sarkar FH. Exploitation of the Notch signaling pathway as a novel target for cancer therapy. Anticancer Res. 2008;28:3621-3630.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Lobry C, Oh P, Aifantis I. Oncogenic and tumor suppressor functions of Notch in cancer: it’s NOTCH what you think. J Exp Med. 2011;208:1931-1935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 276]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
87.  Radtke F, Raj K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nat Rev Cancer. 2003;3:756-767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 600]  [Cited by in F6Publishing: 596]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
88.  Smith DC, Eisenberg PD, Manikhas G, Chugh R, Gubens MA, Stagg RJ, Kapoun AM, Xu L, Dupont J, Sikic B. A phase I dose escalation and expansion study of the anticancer stem cell agent demcizumab (anti-DLL4) in patients with previously treated solid tumors. Clin Cancer Res. 2014;20:6295-6303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 115]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
89.  Gracian AC, Jameson MB, Grande E, Cooray P, Parnis F, Grimison P, Jeffery M, Stagg RJ, Dupont J, Tebbutt NC. A phase 1b study of the anticancer stem cell agent demcizumab (dem) and gemcitabine (gem) with or without paclitaxel protein bound particles (nab-paclitaxel) in patients with pancreatic cancer. J Clin Oncol. 2014;32 Suppl 3:Abstr 279.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  McKeage MJ, Kotasek D, Markman B, Hidalgo M, Millward M, Jameson MB, Harris DL, Stagg RJ, Dupont J, Hughes BGM. A phase 1b study of the anticancer stem cell agent demcizumab (dem), pemetrexed (pem), and carboplatin (carbo) in pts with first-line nonsquamous NSCLC. J Clin Oncol. 2014;32 Suppl 3:Abstr 2544.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4035]  [Cited by in F6Publishing: 4332]  [Article Influence: 393.8]  [Reference Citation Analysis (0)]
92.  Dontu G, Jackson KW, McNicholas E, Kawamura MJ, Abdallah WM, Wicha MS. Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells. Breast Cancer Res. 2004;6:R605-R615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 556]  [Cited by in F6Publishing: 589]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
93.  Harrison H, Farnie G, Howell SJ, Rock RE, Stylianou S, Brennan KR, Bundred NJ, Clarke RB. Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Cancer Res. 2010;70:709-718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 387]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
94.  Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, Krop I, Paskett LA, Wong H, Dobrolecki LE, Lewis MT. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19:1512-1524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 186]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
95.  Krop I, Demuth T, Guthrie T, Wen PY, Mason WP, Chinnaiyan P, Butowski N, Groves MD, Kesari S, Freedman SJ. Phase I pharmacologic and pharmacodynamic study of the gamma secretase (Notch) inhibitor MK-0752 in adult patients with advanced solid tumors. J Clin Oncol. 2012;30:2307-2313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 236]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
96.  Eniu A, Palmieri FM, Perez EA. Weekly administration of docetaxel and paclitaxel in metastatic or advanced breast cancer. Oncologist. 2005;10:665-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
97.  Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nat Rev Cancer. 2008;8:387-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1156]  [Cited by in F6Publishing: 1167]  [Article Influence: 72.9]  [Reference Citation Analysis (0)]
98.  Wend P, Holland JD, Ziebold U, Birchmeier W. Wnt signaling in stem and cancer stem cells. Semin Cell Dev Biol. 2010;21:855-863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 151]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
99.  Katoh M, Katoh M. WNT signaling pathway and stem cell signaling network. Clin Cancer Res. 2007;13:4042-4045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 519]  [Cited by in F6Publishing: 569]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
100.  Gurney A, Axelrod F, Bond CJ, Cain J, Chartier C, Donigan L, Fischer M, Chaudhari A, Ji M, Kapoun AM. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc Natl Acad Sci USA. 2012;109:11717-11722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 378]  [Cited by in F6Publishing: 436]  [Article Influence: 36.3]  [Reference Citation Analysis (0)]
101.  Smith DC, Rosen LS, Chugh R, Goldman JW, Xu L, Kapoun A, Brachmann RK, Dupont J, Stagg RJ, Tolcher AW. First-in-human evaluation of the human monoclonal antibody vantictumab (OMP-18R5; anti-frizzled) targeting the Wnt pathway in a phase I study for patients with advanced solid tumors. J Clin Oncol. 2013;31:abstr 2540.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Jimeno A, Gordon MS, Chugh R, Messersmith WA, Mendelson DS, Dupont J, Stagg RJ, Kapoun A, Xu L, Brachmann RK. A first-in-human phase 1 study of anticancer stem cell agent OMP-54F28 (fzd8-fc), decoy receptor for wnt ligands, in patients with advanced solid tumors. J Clin Oncol. 2014;32 Suppl 3:Abstr 2505.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol. 2000;18:217-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1803]  [Cited by in F6Publishing: 1759]  [Article Influence: 73.3]  [Reference Citation Analysis (0)]
104.  Walz A, Peveri P, Aschauer H, Baggiolini M. Purification and amino acid sequencing of NAF, a novel neutrophil-activating factor produced by monocytes. Biochem Biophys Res Commun. 1987;149:755-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 348]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
105.  Schröder JM, Mrowietz U, Morita E, Christophers E. Purification and partial biochemical characterization of a human monocyte-derived, neutrophil-activating peptide that lacks interleukin 1 activity. J Immunol. 1987;139:3474-3483.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Holmes WE, Lee J, Kuang WJ, Rice GC, Wood WI. Structure and functional expression of a human interleukin-8 receptor. Science. 1991;253:1278-1280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 791]  [Cited by in F6Publishing: 781]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
107.  Murphy PM, Tiffany HL. Cloning of complementary DNA encoding a functional human interleukin-8 receptor. Science. 1991;253:1280-1283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 662]  [Cited by in F6Publishing: 649]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
108.  Venkatakrishnan G, Salgia R, Groopman JE. Chemokine receptors CXCR-1/2 activate mitogen-activated protein kinase via the epidermal growth factor receptor in ovarian cancer cells. J Biol Chem. 2000;275:6868-6875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 171]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
109.  Zhu YM, Webster SJ, Flower D, Woll PJ. Interleukin-8/CXCL8 is a growth factor for human lung cancer cells. Br J Cancer. 2004;91:1970-1976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 176]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
110.  Singh JK, Simões BM, Clarke RB, Bundred NJ. Targeting IL-8 signalling to inhibit breast cancer stem cell activity. Expert Opin Ther Targets. 2013;17:1235-1241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
111.  Bertini R, Allegretti M, Bizzarri C, Moriconi A, Locati M, Zampella G, Cervellera MN, Di Cioccio V, Cesta MC, Galliera E. Noncompetitive allosteric inhibitors of the inflammatory chemokine receptors CXCR1 and CXCR2: prevention of reperfusion injury. Proc Natl Acad Sci USA. 2004;101:11791-11796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 260]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
112.  Citro A, Cantarelli E, Maffi P, Nano R, Melzi R, Mercalli A, Dugnani E, Sordi V, Magistretti P, Daffonchio L. CXCR1/2 inhibition enhances pancreatic islet survival after transplantation. J Clin Invest. 2012;122:3647-3651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
113.  Schott A, Wicha M, Cristofanilli M, Ruffini P, McCanna S, Reuben J, Goldstein L. Abstract ot2-3-01: Phase Ib pilot study to evaluate reparixin in combination with chemotherapy with weekly paclitaxel in patients with HER-2 negative metastatic breast cancer (MBC). Cancer Res. 2012;72:OT2-3-01.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Goldstein L, Sparano J, Perez R, Vito C, Reuben J, Landis M, McCanna S, Ruffini P, Cristofanilli M, Chang J. A single arm, preoperative, pilot study to evaluate the safety and biological effects of orally administered reparixin in early breast cancer patients who are candidates for surgery. Cancer Res. 2013;73:OT2-6-03.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Singh JK, Farnie G, Bundred NJ, Simões BM, Shergill A, Landberg G, Howell SJ, Clarke RB. Targeting CXCR1/2 significantly reduces breast cancer stem cell activity and increases the efficacy of inhibiting HER2 via HER2-dependent and -independent mechanisms. Clin Cancer Res. 2013;19:643-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 170]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
116.  Marotta LL, Almendro V, Marusyk A, Shipitsin M, Schemme J, Walker SR, Bloushtain-Qimron N, Kim JJ, Choudhury SA, Maruyama R. The JAK2/STAT3 signaling pathway is required for growth of CD44+CD24- stem cell-like breast cancer cells in human tumors. J Clin Invest. 2011;121:2723-2735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 654]  [Cited by in F6Publishing: 701]  [Article Influence: 53.9]  [Reference Citation Analysis (0)]
117.  Birnbaum T, Roider J, Schankin CJ, Padovan CS, Schichor C, Goldbrunner R, Straube A. Malignant gliomas actively recruit bone marrow stromal cells by secreting angiogenic cytokines. J Neurooncol. 2007;83:241-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 138]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
118.  Waugh DJ, Wilson C. The interleukin-8 pathway in cancer. Clin Cancer Res. 2008;14:6735-6741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1408]  [Cited by in F6Publishing: 1522]  [Article Influence: 95.1]  [Reference Citation Analysis (0)]
119.  Korkaya H, Kim GI, Davis A, Malik F, Henry NL, Ithimakin S, Quraishi AA, Tawakkol N, D’Angelo R, Paulson AK. Activation of an IL6 inflammatory loop mediates trastuzumab resistance in HER2+ breast cancer by expanding the cancer stem cell population. Mol Cell. 2012;47:570-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 409]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
120.  Korkaya H, Paulson A, Iovino F, Wicha MS. HER2 regulates the mammary stem/progenitor cell population driving tumorigenesis and invasion. Oncogene. 2008;27:6120-6130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 414]  [Cited by in F6Publishing: 414]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
121.  Magnifico A, Albano L, Campaner S, Delia D, Castiglioni F, Gasparini P, Sozzi G, Fontanella E, Menard S, Tagliabue E. Tumor-initiating cells of HER2-positive carcinoma cell lines express the highest oncoprotein levels and are sensitive to trastuzumab. Clin Cancer Res. 2009;15:2010-2021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 206]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
122.  Korkaya H, Paulson A, Charafe-Jauffret E, Ginestier C, Brown M, Dutcher J, Clouthier SG, Wicha MS. Regulation of mammary stem/progenitor cells by PTEN/Akt/beta-catenin signaling. PLoS Biol. 2009;7:e1000121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 404]  [Article Influence: 26.9]  [Reference Citation Analysis (0)]
123.  Korkaya H, Wicha MS. HER2 and breast cancer stem cells: more than meets the eye. Cancer Res. 2013;73:3489-3493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 93]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
124.  Paik S, Kim C, Wolmark N. HER2 status and benefit from adjuvant trastuzumab in breast cancer. N Engl J Med. 2008;358:1409-1411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 336]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
125.  Ithimakin S, Day KC, Malik F, Zen Q, Dawsey SJ, Bersano-Begey TF, Quraishi AA, Ignatoski KW, Daignault S, Davis A. HER2 drives luminal breast cancer stem cells in the absence of HER2 amplification: implications for efficacy of adjuvant trastuzumab. Cancer Res. 2013;73:1635-1646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 193]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
126.  Korkaya H, Wicha MS. Breast cancer stem cells: we’ve got them surrounded. Clin Cancer Res. 2013;19:511-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
127.  Paller CJ, Bradbury PA, Ivy SP, Seymour L, LoRusso PM, Baker L, Rubinstein L, Huang E, Collyar D, Groshen S. Design of phase I combination trials: recommendations of the Clinical Trial Design Task Force of the NCI Investigational Drug Steering Committee. Clin Cancer Res. 2014;20:4210-4217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
128.  Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, Lander ES. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell. 2009;138:645-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1786]  [Cited by in F6Publishing: 1832]  [Article Influence: 122.1]  [Reference Citation Analysis (0)]
129.  Sachlos E, Risueño RM, Laronde S, Shapovalova Z, Lee JH, Russell J, Malig M, McNicol JD, Fiebig-Comyn A, Graham M. Identification of drugs including a dopamine receptor antagonist that selectively target cancer stem cells. Cell. 2012;149:1284-1297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 359]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
130.  Janku F, McConkey DJ, Hong DS, Kurzrock R. Autophagy as a target for anticancer therapy. Nat Rev Clin Oncol. 2011;8:528-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 588]  [Cited by in F6Publishing: 618]  [Article Influence: 47.5]  [Reference Citation Analysis (0)]
131.  Gong C, Bauvy C, Tonelli G, Yue W, Deloménie C, Nicolas V, Zhu Y, Domergue V, Marin-Esteban V, Tharinger H. Beclin 1 and autophagy are required for the tumorigenicity of breast cancer stem-like/progenitor cells. Oncogene. 2013;32:2261-2272, 2272e.1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 260]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
132.  Bellodi C, Lidonnici MR, Hamilton A, Helgason GV, Soliera AR, Ronchetti M, Galavotti S, Young KW, Selmi T, Yacobi R. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin Invest. 2009;119:1109-1123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 450]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
133.  Kourelis TV, Siegel RD. Metformin and cancer: new applications for an old drug. Med Oncol. 2012;29:1314-1327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 217]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
134.  Rattan R, Ali Fehmi R, Munkarah A. Metformin: an emerging new therapeutic option for targeting cancer stem cells and metastasis. J Oncol. 2012;2012:928127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 83]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
135.  Song CW, Lee H, Dings RP, Williams B, Powers J, Santos TD, Choi BH, Park HJ. Metformin kills and radiosensitizes cancer cells and preferentially kills cancer stem cells. Sci Rep. 2012;2:362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 212]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
136.  Thompson AM. Molecular pathways: preclinical models and clinical trials with metformin in breast cancer. Clin Cancer Res. 2014;20:2508-2515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
137.  Kim J, Tang JY, Gong R, Kim J, Lee JJ, Clemons KV, Chong CR, Chang KS, Fereshteh M, Gardner D. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell. 2010;17:388-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 377]  [Cited by in F6Publishing: 379]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
138.  Kim J, Lee JJ, Kim J, Gardner D, Beachy PA. Arsenic antagonizes the Hedgehog pathway by preventing ciliary accumulation and reducing stability of the Gli2 transcriptional effector. Proc Natl Acad Sci USA. 2010;107:13432-13437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 246]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
139.  Kim DJ, Kim J, Spaunhurst K, Montoya J, Khodosh R, Chandra K, Fu T, Gilliam A, Molgo M, Beachy PA. Open-label, exploratory phase II trial of oral itraconazole for the treatment of basal cell carcinoma. J Clin Oncol. 2014;32:745-751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 184]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
140.  Bhat-Nakshatri P, Goswami CP, Badve S, Sledge GW, Nakshatri H. Identification of FDA-approved drugs targeting breast cancer stem cells along with biomarkers of sensitivity. Sci Rep. 2013;3:2530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
141.  Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15860]  [Cited by in F6Publishing: 19008]  [Article Influence: 1267.2]  [Reference Citation Analysis (1)]
142.  Steeg PS. Perspective: The right trials. Nature. 2012;485:S58-S59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
143.  Oxnard GR, Morris MJ, Hodi FS, Baker LH, Kris MG, Venook AP, Schwartz LH. When progressive disease does not mean treatment failure: reconsidering the criteria for progression. J Natl Cancer Inst. 2012;104:1534-1541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 103]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
144.  Oxnard GR, Arcila ME, Chmielecki J, Ladanyi M, Miller VA, Pao W. New strategies in overcoming acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in lung cancer. Clin Cancer Res. 2011;17:5530-5537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 285]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
145.  Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W, Ferrari D, Chen Y, Costa DB, Gerber DE, Orlov S. Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011;29:3307-3315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 343]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
146.  Giuliano M, Giordano A, Jackson S, De Giorgi U, Mego M, Cohen EN, Gao H, Anfossi S, Handy BC, Ueno NT. Circulating tumor cells as early predictors of metastatic spread in breast cancer patients with limited metastatic dissemination. Breast Cancer Res. 2014;16:440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 87]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
147.  Smith MR, Saad F, Coleman R, Shore N, Fizazi K, Tombal B, Miller K, Sieber P, Karsh L, Damião R. Denosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase 3, randomised, placebo-controlled trial. Lancet. 2012;379:39-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 589]  [Cited by in F6Publishing: 571]  [Article Influence: 47.6]  [Reference Citation Analysis (0)]
148.  Stopeck AT, Lipton A, Body JJ, Steger GG, Tonkin K, de Boer RH, Lichinitser M, Fujiwara Y, Yardley DA, Viniegra M. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol. 2010;28:5132-5139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1096]  [Cited by in F6Publishing: 1018]  [Article Influence: 72.7]  [Reference Citation Analysis (0)]
149.  Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3360]  [Cited by in F6Publishing: 3257]  [Article Influence: 162.9]  [Reference Citation Analysis (0)]
150.  Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, Matera J, Allard WJ, Doyle GV, Terstappen LW. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. 2006;12:4218-4224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 765]  [Cited by in F6Publishing: 752]  [Article Influence: 44.2]  [Reference Citation Analysis (0)]
151.  Haber DA, Velculescu VE. Blood-based analyses of cancer: circulating tumor cells and circulating tumor DNA. Cancer Discov. 2014;4:650-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 507]  [Article Influence: 50.7]  [Reference Citation Analysis (0)]
152.  Grover PK, Cummins AG, Price TJ, Roberts-Thomson IC, Hardingham JE. Circulating tumour cells: the evolving concept and the inadequacy of their enrichment by EpCAM-based methodology for basic and clinical cancer research. Ann Oncol. 2014;25:1506-1516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 158]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
153.  Dave B, Granados-Principal S, Zhu R, Benz S, Rabizadeh S, Soon-Shiong P, Yu KD, Shao Z, Li X, Gilcrease M. Targeting RPL39 and MLF2 reduces tumor initiation and metastasis in breast cancer by inhibiting nitric oxide synthase signaling. Proc Natl Acad Sci USA. 2014;111:8838-8843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 88]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
154.  Ellis LM, Bernstein DS, Voest EE, Berlin JD, Sargent D, Cortazar P, Garrett-Mayer E, Herbst RS, Lilenbaum RC, Sima C. American Society of Clinical Oncology perspective: Raising the bar for clinical trials by defining clinically meaningful outcomes. J Clin Oncol. 2014;32:1277-1280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 301]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
155.  Bonadonna G, Moliterni A, Zambetti M, Daidone MG, Pilotti S, Gianni L, Valagussa P. 30 years’ follow up of randomised studies of adjuvant CMF in operable breast cancer: cohort study. BMJ. 2005;330:217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 206]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
156.  Yauch RL, Dijkgraaf GJ, Alicke B, Januario T, Ahn CP, Holcomb T, Pujara K, Stinson J, Callahan CA, Tang T. Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science. 2009;326:572-574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 658]  [Cited by in F6Publishing: 670]  [Article Influence: 44.7]  [Reference Citation Analysis (0)]
157.  Atwood SX, Li M, Lee A, Tang JY, Oro AE. GLI activation by atypical protein kinase C ι/λ regulates the growth of basal cell carcinomas. Nature. 2013;494:484-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 192]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]